Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 82
Filtrar
1.
PLoS Negl Trop Dis ; 8(10): e3268, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25329054

RESUMO

We have previously reported that Vivax Malaria Protein 001 (VMP001), a vaccine candidate based on the circumsporozoite protein of Plasmodium vivax, is immunogenic in mice and rhesus monkeys in the presence of various adjuvants. In the present study, we evaluated the immunogenicity and efficacy of VMP001 formulated with a TLR9 agonist in a water-in-oil emulsion. Following immunization, the vaccine efficacy was assessed by challenging Aotus nancymaae monkeys with P. vivax sporozoites. Monkeys from both the low- and high-dose vaccine groups generated strong humoral immune responses to the vaccine (peak median titers of 291,622), and its subunits (peak median titers to the N-term, central repeat and C-term regions of 22,188; 66,120 and 179,947, respectively). 66.7% of vaccinated monkeys demonstrated sterile protection following challenge. Protection was associated with antibodies directed against the central repeat region. The protected monkeys had a median anti-repeat titer of 97,841 compared to 14,822 in the non-protected monkeys. This is the first report demonstrating P. vivax CSP vaccine-induced protection of Aotus monkeys challenged with P. vivax sporozoites.


Assuntos
Aotidae/imunologia , Vacinas Antimaláricas/imunologia , Malária Vivax/imunologia , Plasmodium vivax/imunologia , Proteínas de Protozoários/imunologia , Adjuvantes Imunológicos , Animais , Anticorpos Antiprotozoários/sangue , Anticorpos Antiprotozoários/imunologia , Aotidae/parasitologia , Feminino , Imunidade Humoral/imunologia , Malária Vivax/parasitologia , Malária Vivax/prevenção & controle , Masculino , Camundongos , Doenças dos Macacos/parasitologia , Doenças dos Macacos/prevenção & controle , Distribuição Aleatória , Receptor Toll-Like 9/imunologia , Vacinação , Vacinas Sintéticas/imunologia
2.
Infect Genet Evol ; 20: 239-48, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24044894

RESUMO

The merozoite surface protein-9 (MSP-9) has been considered a target for an anti-malarial vaccine since it is one of many proteins involved in the erythrocyte invasion, a critical step in the parasite life cycle. Orthologs encoding this antigen have been found in all known species of Plasmodium parasitic to primates. In order to characterize and investigate the extent and maintenance of MSP-9 genetic diversity, we analyzed DNA sequences of the following malaria parasite species: Plasmodium falciparum, Plasmodium reichenowi, Plasmodium chabaudi, Plasmodium yoelii, Plasmodium berghei, Plasmodium coatneyi, Plasmodium gonderi, Plasmodium knowlesi, Plasmodium inui, Plasmodium simiovale, Plasmodium fieldi, Plasmodium cynomolgi and Plasmodium vivax and evaluated the signature of natural selection in all MSP-9 orthologs. Our findings suggest that the gene encoding MSP-9 is under purifying selection in P. vivax and closely related species. We further explored how selection affected different regions of MSP-9 by comparing the polymorphisms in P. vivax and P. falciparum, and found contrasting patterns between these two species that suggest differences in functional constraints. This observation implies that the MSP-9 orthologs in human parasites may interact differently with the host immune response. Thus, studies carried out in one species cannot be directly translated into the other.


Assuntos
Vacinas Antimaláricas/imunologia , Malária Vivax/imunologia , Proteínas de Membrana/genética , Plasmodium vivax/imunologia , Proteínas de Protozoários/genética , Antígenos de Protozoários/genética , Antígenos de Protozoários/imunologia , Sequência de Bases , Clonagem Molecular , Epitopos de Linfócito T/genética , Epitopos de Linfócito T/imunologia , Variação Genética , Humanos , Vacinas Antimaláricas/genética , Malária Vivax/prevenção & controle , Proteínas de Membrana/imunologia , Filogenia , Plasmodium falciparum/genética , Plasmodium falciparum/imunologia , Plasmodium vivax/genética , Polimorfismo de Nucleotídeo Único , Proteínas de Protozoários/imunologia , Alinhamento de Sequência , Análise de Sequência de DNA
3.
J Infect Dis ; 208(2): 260-70, 2013 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-23568175

RESUMO

Whether vaccination against a virus can protect against more virulent coinfection with the virus and additional pathogen(s) remains poorly characterized. Overlapping endemicity of human immunodeficiency virus (HIV) and malaria suggests that HIV/malaria coinfection frequently complicates acute and chronic HIV infection. Here we showed that vaccination of macaques with recombinant Listeria ΔactA prfA* expressing simian/human immunodeficiency virus (SHIV) gag and env elicited Gag- and Env-specific T-cell responses, and protected against life-threatening SHIV-related malaria after SHIV/Plasmodium fragile coinfection. SHIV antigen immunization reduced peak viremia, resisted SHIV/malaria-induced lymphoid destruction, and blunted coinfection-accelerated decline of CD4(+) T-cell counts after SHIV/malaria coinfection. SHIV antigen immunization also weakened coinfection-driven overreactive proinflammatory interferon-γ (IFNγ) responses and led to developing T helper cell 17/22 (Th17/Th22) responses after SHIV/malaria coinfection. The findings suggest that vaccination against AIDS virus can alter patterns of immune responses to the SHIV/malaria coinfection and protect against life-threatening SHIV-related malaria.


Assuntos
Antígenos Virais/imunologia , Coinfecção/imunologia , Infecções por HIV/imunologia , Malária/imunologia , Síndrome de Imunodeficiência Adquirida dos Símios/imunologia , Vírus da Imunodeficiência Símia/imunologia , Animais , Linfócitos T CD4-Positivos/imunologia , Coinfecção/microbiologia , Coinfecção/parasitologia , Coinfecção/prevenção & controle , Produtos do Gene env/imunologia , Produtos do Gene gag/imunologia , Infecções por HIV/parasitologia , Infecções por HIV/prevenção & controle , Infecções por HIV/virologia , HIV-1/imunologia , Macaca mulatta/imunologia , Macaca mulatta/microbiologia , Macaca mulatta/virologia , Malária/microbiologia , Malária/prevenção & controle , Plasmodium/imunologia , Síndrome de Imunodeficiência Adquirida dos Símios/prevenção & controle , Síndrome de Imunodeficiência Adquirida dos Símios/virologia , Linfócitos T Auxiliares-Indutores/imunologia , Vacinação/métodos , Vacinas Sintéticas/imunologia
4.
Adv Parasitol ; 81: 77-131, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23384622

RESUMO

Population studies show that individuals acquire immunity to Plasmodium vivax more quickly than Plasmodium falciparum irrespective of overall transmission intensity, resulting in the peak burden of P. vivax malaria in younger age groups. Similarly, actively induced P. vivax infections in malaria therapy patients resulted in faster and generally more strain-transcending acquisition of immunity than P. falciparum infections. The mechanisms behind the more rapid acquisition of immunity to P. vivax are poorly understood. Natural acquired immune responses to P. vivax target both pre-erythrocytic and blood-stage antigens and include humoral and cellular components. To date, only a few studies have investigated the association of these immune responses with protection, with most studies focussing on a few merozoite antigens (such as the Pv Duffy binding protein (PvDBP), the Pv reticulocyte binding proteins (PvRBPs), or the Pv merozoite surface proteins (PvMSP1, 3 & 9)) or the circumsporozoite protein (PvCSP). Naturally acquired transmission-blocking (TB) immunity (TBI) was also found in several populations. Although limited, these data support the premise that developing a multi-stage P. vivax vaccine may be feasible and is worth pursuing.


Assuntos
Imunidade Adaptativa/imunologia , Malária Vivax/imunologia , Plasmodium vivax/imunologia , Animais , Antígenos de Protozoários/imunologia , Humanos , Malária Falciparum/epidemiologia , Malária Falciparum/imunologia , Malária Vivax/epidemiologia , Plasmodium falciparum/imunologia
5.
Infect Genet Evol ; 12(5): 978-86, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22414917

RESUMO

Evidence for natural selection, positive or negative, on gene encoding antigens may indicate variation or functional constraints that are immunologically relevant. Most malaria surface antigens with high genetic diversity have been reported to be under positive-diversifying selection. However, antigens with limited genetic variation are usually ignored in terms of the role that natural selection may have in generating such patterns. We investigated orthologous genes encoding two merozoite proteins, MSP8 and MSP10, among several mammalian Plasmodium spp. These antigens, together with MSP1, are among the few MSPs that have two epidermal growth factor-like domains (EGF) at the C-terminal. Those EGF are relatively conserved (low levels of genetic polymorphism) and have been proposed to act as ligands during the invasion of RBCs. We use several evolutionary genetic methods to detect patterns consistent with natural selection acting on MSP8 and MSP10 orthologs in the human parasites Plasmodium falciparum and P. vivax, as well as closely related malarial species found in non-human primates (NHPs). Overall, these antigens have low polymorphism in the human parasites in comparison with the orthologs from other Plasmodium spp. We found that the MSP10 gene polymorphism in P. falciparum only harbor non-synonymous substitutions, a pattern consistent with a gene under positive selection. Evidence of purifying selection was found on the polymorphism observed in both orthologs from P. cynomolgi, a non-human primate parasite closely related to P. vivax, but it was not conclusive in the human parasite. Yet, using phylogenetic base approaches, we found evidence for purifying selection on both MSP8 and MSP10 in the lineage leading to P. vivax. Such antigens evolving under strong functional constraints could become valuable vaccine candidates. We discuss how comparative approaches could allow detecting patterns consistent with negative selection even when there is low polymorphism in the extant populations.


Assuntos
Antígenos de Protozoários/genética , Plasmodium/genética , Polimorfismo Genético/genética , Proteínas de Protozoários/genética , Sequência de Aminoácidos , Antígenos de Protozoários/química , Teorema de Bayes , Sítios de Ligação/genética , Fator de Crescimento Epidérmico/química , Evolução Molecular , Genes de Protozoários/genética , Humanos , Malária/parasitologia , Dados de Sequência Molecular , Filogenia , Plasmodium/química , Estrutura Terciária de Proteína/genética , Proteínas de Protozoários/química , Seleção Genética , Alinhamento de Sequência
6.
Am J Trop Med Hyg ; 86(3): 398-402, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22403307

RESUMO

Oocyst counts were compared between mosquitoes that fed on humans versus mosquitoes that fed on Aotus monkeys, both of which were infected with the Chesson strain of Plasmodium vivax. Oocyst counts obtained from mosquitoes fed on humans were almost 10-fold higher in number. Mosquitoes were more likely to be infected and with a higher rate of infection when they fed on monkeys before the peak in the asexual parasite count. Mosquitoes that fed on humans were more likely to be more heavily infected when fed after the peak in the asexual count. Of several species of owl monkeys, Aotus vociferans was infected at a higher frequency. On the basis of oocyst counts, Anopheles dirus were the most susceptible and An. maculatus were the least susceptible of the mosquito species tested.


Assuntos
Anopheles/parasitologia , Aotidae/parasitologia , Plasmodium vivax/patogenicidade , Animais , Antimaláricos/uso terapêutico , Cloroquina/uso terapêutico , Gerenciamento Clínico , Modelos Animais de Doenças , Interações Hospedeiro-Parasita , Humanos , Malária/parasitologia , Malária/veterinária , Masculino , Doenças dos Macacos/parasitologia , Primaquina/uso terapêutico , Proguanil/uso terapêutico , Pirimetamina/uso terapêutico , Quinina/uso terapêutico
7.
Annu Rev Entomol ; 57: 107-21, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22149265

RESUMO

Plasmodium knowlesi is a malaria parasite of monkeys of Southeast Asia that is transmitted by mosquitoes of the Anopheles leucosphyrus group. Humans are frequently infected with this parasite and misdiagnosed as being infected with Plasmodium malariae. The parasite was a major monkey animal model for developing antimalarial vaccines and investigations of the biology of parasite invasion. P. knowlesi is the first monkey malaria parasite genome to be sequenced and annotated.


Assuntos
Malária/parasitologia , Doenças dos Macacos/parasitologia , Plasmodium knowlesi/fisiologia , Animais , Haplorrinos/parasitologia , Interações Hospedeiro-Parasita , Humanos , Insetos Vetores/parasitologia , Malária/imunologia , Malária/transmissão
8.
J Infect Dis ; 204(9): 1450-62, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21921207

RESUMO

BACKGROUND: Malaria and AIDS represent 2 leading causes of death from infectious diseases worldwide, and their high geographic overlap means coinfection is prevalent. It remains unknown whether distinct immune responses during coinfection with malaria and human immunodeficiency virus (HIV) affect clinical outcomes. METHODS: We tested this hypothesis by employing macaque models of coinfection with malaria and simian-human immunodeficiency virus (SHIV). RESULTS: Plasmodium fragile malaria coinfection of acutely SHIV-infected macaques induced hyperimmune activation and remarkable expansion of CD4+ and CD8+ T effector cells de novo producing interferon γ or tumor necrosis factor α. Malaria-driven cellular hyperactivation/expansion and high-level Th1-cytokines enhanced SHIV disease characterized by increasing CD4+ T-cell depletion, profound lymphoid depletion or destruction, and even necrosis in lymph nodes and spleens. Importantly, malaria/SHIV-mediated depletion, destruction, and necrosis in lymphoid tissues led to bursting parasite replication and fatal virus-associated malaria. Surprisingly, chronically SHIV-infected macaques without AIDS employed different defense mechanisms during malaria coinfection, and mounted unique ∼200-fold expansion of interleukin 17+/interleukin 22+ T effectors with profound Th1 suppression. Such remarkable expansion of Th17/Th22 cells and inhibition of Th1 response coincided with development of immunity against fatal virus-associated malaria without accelerating SHIV disease. CONCLUSIONS: These novel findings suggest that virus infection status and selected Th1 or Th17/Th22 responses after malaria/AIDS-virus coinfection correlate with distinct outcomes of virus infection and malaria.


Assuntos
Malária/imunologia , Síndrome de Imunodeficiência Adquirida dos Símios/imunologia , Células Th1/imunologia , Células Th17/imunologia , Animais , Modelos Animais de Doenças , Macaca mulatta , Plasmodium/imunologia , Plasmodium/patogenicidade , Doenças dos Primatas/imunologia , Vírus da Imunodeficiência Símia/imunologia , Vírus da Imunodeficiência Símia/patogenicidade , Resultado do Tratamento
9.
J Virol ; 85(22): 11655-63, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21917966

RESUMO

In sub-Saharan Africa, HIV-1 infection frequently occurs in the context of other coinfecting pathogens, most importantly, Mycobacterium tuberculosis and malaria parasites. The consequences are often devastating, resulting in enhanced morbidity and mortality. Due to the large number of confounding factors influencing pathogenesis in coinfected people, we sought to develop a nonhuman primate model of simian immunodeficiency virus (SIV)-malaria coinfection. In sub-Saharan Africa, Plasmodium falciparum is the most common malaria parasite and is responsible for most malaria-induced deaths. The simian malaria parasite Plasmodium fragile can induce clinical symptoms, including cerebral malaria in rhesus macaques, that resemble those of P. falciparum infection in humans. Thus, based on the well-characterized rhesus macaque model of SIV infection, this study reports the development of a novel rhesus macaque SIV-P. fragile coinfection model to study human HIV-P. falciparum coinfection. Using this model, we show that coinfection is associated with an increased, although transient, risk of both HIV and malaria transmission. Specifically, SIV-P. fragile coinfected macaques experienced an increase in SIV viremia that was temporarily associated with an increase in potential SIV target cells and systemic immune activation during acute parasitemia. Conversely, primary parasitemia in SIV-P. fragile coinfected animals resulted in higher gametocytemia that subsequently translated into higher oocyst development in mosquitoes. To our knowledge, this is the first animal model able to recapitulate the increased transmission risk of both HIV and malaria in coinfected humans. Therefore, this model could serve as an essential tool to elucidate distinct immunological, virological, and/or parasitological parameters underlying disease exacerbation in HIV-malaria coinfected people.


Assuntos
Malária/complicações , Malária/transmissão , Plasmodium falciparum/patogenicidade , Síndrome de Imunodeficiência Adquirida dos Símios/complicações , Síndrome de Imunodeficiência Adquirida dos Símios/transmissão , Vírus da Imunodeficiência Símia/patogenicidade , Animais , Modelos Animais de Doenças , Humanos , Macaca mulatta , Masculino , Parasitemia/complicações , Plasmodium falciparum/isolamento & purificação , Doenças dos Primatas/transmissão , Vírus da Imunodeficiência Símia/isolamento & purificação , Viremia/complicações
10.
PLoS One ; 5(12): e14275, 2010 Dec 09.
Artigo em Inglês | MEDLINE | ID: mdl-21151554

RESUMO

Plasmodium vivax (Pv) is the second most important human malaria parasite. Recent data indicate that the impact of Pv malaria on the health and economies of the developing world has been dramatically underestimated. Pv has a unique feature in its life cycle. Uninucleate sporozoites (spz), after invasion of human hepatocytes, either proceed to develop into tens of thousands of merozoites within the infected hepatocytes or remain as dormant forms called hypnozoites, which cause relapses of malaria months to several years after the primary infection. Elimination of malaria caused by Pv will be facilitated by developing a safe, highly effective drug that eliminates Pv liver stages, including hypnozoites. Identification and development of such a drug would be facilitated by the development of a medium to high throughput assay for screening drugs against Pv liver stages. We undertook the present pilot study to (1) assess the feasibility of producing large quantities of purified, vialed, cryopreserved Pv sporozoites and (2) establish a system for culturing the liver stages of Pv in order to assess the effects of drugs on the liver stages of Pv. We used primaquine (PQ) to establish this assay model, because PQ is the only licensed drug known to clear all Pv hepatocyte stages, including hypnozoites, and the effect of PQ on Pv hepatocyte stage development in vitro has not previously been reported. We report that we have established the capacity to reproducibly infect hepatoma cells with purified, cyropreserved Pv spz from the same lot, quantitate the primary outcome variable of infected hepatoma cells and demonstrate the inhibitory activity of primaquine on the infected hepatoma cells. We have also identified small parasite forms that may be hypnozoites. These data provide the foundation for finalizing a medium throughput, high content assay to identify new drugs for the elimination of all Pv liver stages.


Assuntos
Fígado/efeitos dos fármacos , Fígado/metabolismo , Malária Vivax/metabolismo , Animais , Anopheles/parasitologia , Antimaláricos/farmacologia , Carcinoma Hepatocelular/tratamento farmacológico , Carcinoma Hepatocelular/parasitologia , Criopreservação , Modelos Animais de Doenças , Hepatócitos/efeitos dos fármacos , Hepatócitos/parasitologia , Humanos , Técnicas In Vitro , Microscopia de Fluorescência/métodos , Pan troglodytes , Plasmodium vivax/metabolismo , Primaquina/farmacologia , Glândulas Salivares/metabolismo , Esporozoítos/química
11.
Clin Vaccine Immunol ; 17(10): 1631-8, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20702658

RESUMO

Approximately 3.2 billion people live in areas where malaria is endemic, and WHO estimates that 350 to 500 million malaria cases occur each year worldwide. This high prevalence, and the high frequency of international travel, creates significant risk for the exportation of malaria to countries where malaria is not endemic and for the introduction of malaria organisms into the blood supply. Since all four human infectious Plasmodium species have been transmitted by blood transfusion, we sought to develop an enzyme-linked immunosorbent assay (ELISA) capable of detecting antibodies elicited by infection with any of these species. The merozoite surface protein 1 (MSP1), a P. falciparum and P. vivax vaccine candidate with a well-characterized immune response, was selected for use in the assay. The MSP1 genes from P. ovale and P. malariae were cloned and sequenced (L. Birkenmeyer, A. S. Muerhoff, G. Dawson, and S. M. Desai, Am. J. Trop. Med. Hyg. 82:996-1003, 2010), and the carboxyl-terminal p19 regions of all four species were expressed in Escherichia coli. Performance results from individual p19 ELISAs were compared to those of a commercial test (Lab 21 Healthcare Malaria enzyme immunoassay [EIA]). The commercial ELISA detected all malaria patients with P. falciparum or P. vivax infections, as did the corresponding species-specific p19 ELISAs. However, the commercial ELISA detected antibodies in 0/2 and 5/8 individuals with P. malariae and P. ovale infections, respectively, while the p19 assays detected 100% of individuals with confirmed P. malariae or P. ovale infections. In experimentally infected nonhuman primates, the use of MSP1-p19 antigens from all four species resulted in the detection of antibodies within 2 to 10 weeks postinfection. Use of MSP1-p19 antigens from all four Plasmodium species in a single immunoassay would provide significantly improved efficacy compared to existing tests.


Assuntos
Anticorpos Antiprotozoários/sangue , Malária/diagnóstico , Proteína 1 de Superfície de Merozoito , Parasitologia/métodos , Animais , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática/métodos , Escherichia coli/genética , Expressão Gênica , Humanos , Proteína 1 de Superfície de Merozoito/genética , Pan troglodytes , Plasmodium falciparum/genética , Plasmodium falciparum/imunologia , Plasmodium malariae/genética , Plasmodium malariae/imunologia , Plasmodium ovale/genética , Plasmodium ovale/imunologia , Plasmodium vivax/genética , Plasmodium vivax/imunologia , Platirrinos , Proteínas Recombinantes/genética , Sensibilidade e Especificidade
12.
PLoS Negl Trop Dis ; 4(4): e653, 2010 Apr 06.
Artigo em Inglês | MEDLINE | ID: mdl-20386602

RESUMO

BACKGROUND: Up to 40% of the world's population is at risk for Plasmodium vivax malaria, a disease that imposes a major public health and economic burden on endemic countries. Because P. vivax produces latent liver forms, eradication of P. vivax malaria is more challenging than it is for P. falciparum. Genetic analysis of P. vivax is exceptionally difficult due to limitations of in vitro culture. To overcome the barriers to traditional molecular biology in P. vivax, we examined parasite transcriptional changes in samples from infected patients and mosquitoes in order to characterize gene function, define regulatory sequences and reveal new potential vaccine candidate genes. PRINCIPAL FINDINGS: We observed dramatic changes in transcript levels for various genes at different lifecycle stages, indicating that development is partially regulated through modulation of mRNA levels. Our data show that genes involved in common biological processes or molecular machinery are co-expressed. We identified DNA sequence motifs upstream of co-expressed genes that are conserved across Plasmodium species that are likely binding sites of proteins that regulate stage-specific transcription. Despite their capacity to form hypnozoites we found that P. vivax sporozoites show stage-specific expression of the same genes needed for hepatocyte invasion and liver stage development in other Plasmodium species. We show that many of the predicted exported proteins and members of multigene families show highly coordinated transcription as well. CONCLUSIONS: We conclude that high-quality gene expression data can be readily obtained directly from patient samples and that many of the same uncharacterized genes that are upregulated in different P. vivax lifecycle stages are also upregulated in similar stages in other Plasmodium species. We also provide numerous examples of how systems biology is a powerful method for determining the likely function of genes in pathogens that are neglected due to experimental intractability.


Assuntos
Culicidae/parasitologia , Perfilação da Expressão Gênica , Estágios do Ciclo de Vida , Malária Vivax/parasitologia , Plasmodium vivax/genética , Plasmodium vivax/patogenicidade , Animais , Humanos
13.
J Parasitol ; 96(2): 329-39, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19891516

RESUMO

Splenectomized Aotus lemurinus griseimembra, A. azarae boliviensis, A. nancymaae, A. vociferans, and Saimiri boliviensis monkeys were infected with the Uganda I/CDC strain of Plasmodium malariae. The maximum parasite counts were lower if the animals had been previously infected with Plasmodium vivax. Mosquito infection was concentrated in the 12 days following the rise in count above 1,000/microl. Mosquito infection and parasite counts were highest with A. l. griseimembra. Anopheles freeborni was more readily infected than An. gambiae, which was more readily infected than An. stephensi. Parasite counts and mosquito infection with P. brasilianum were much higher in S. boliviensis monkeys than with the Uganda I strain of P. malariae in this host, suggesting marked differences between the host-parasite-vector relationships and indicating that P. brasilianum in S. boliviensis monkeys may be a better reflection of the relationship of P. malariae in the human host.


Assuntos
Anopheles/parasitologia , Aotidae/parasitologia , Insetos Vetores/parasitologia , Plasmodium/fisiologia , Saimiri/parasitologia , Animais , Aotidae/imunologia , Modelos Animais de Doenças , Eritrócitos/parasitologia , Interações Hospedeiro-Parasita , Malária/imunologia , Malária/parasitologia , Parasitemia/imunologia , Parasitemia/parasitologia , Plasmodium/classificação , Plasmodium/imunologia , Plasmodium malariae/classificação , Plasmodium malariae/imunologia , Plasmodium malariae/fisiologia , Análise de Regressão , Saimiri/imunologia , Esplenectomia
14.
PLoS One ; 4(12): e8138, 2009 Dec 03.
Artigo em Inglês | MEDLINE | ID: mdl-19997632

RESUMO

A Plasmodium falciparum 3D7 strain Apical Membrane Antigen-1 (AMA1) vaccine, formulated with AS02(A) adjuvant, slowed parasite growth in a recent Phase 1/2a trial, however sterile protection was not observed. We tested this AS02(A), and a Montanide ISA720 (ISA) formulation of 3D7 AMA1 in Aotus monkeys. The 3D7 parasite does not invade Aotus erythrocytes, hence two heterologous strains, FCH/4 and FVO, were used for challenge, FCH/4 AMA1 being more homologous to 3D7 than FVO AMA1. Following three vaccinations, the monkeys were challenged with 50,000 FCH/4 or 10,000 FVO parasites. Three of the six animals in the AMA+ISA group were protected against FCH/4 challenge. One monkey did not become parasitemic, another showed only a short period of low level parasitemia that self-cured, and a third animal showed a delay before exhibiting its parasitemic phase. This is the first protection shown in primates with a recombinant P. falciparum AMA1 without formulation in Freund's complete adjuvant. No animals in the AMA+AS02(A) group were protected, but this group exhibited a trend towards reduced growth rate. A second group of monkeys vaccinated with AMA+ISA vaccine was not protected against FVO challenge, suggesting strain-specificity of AMA1-based protection. Protection against FCH/4 strain correlated with the quantity of induced antibodies, as the protected animals were the only ones to have in vitro parasite growth inhibitory activity of >70% at 1:10 serum dilution; immuno-fluorescence titers >8,000; ELISA titers against full-length AMA1 >300,000 and ELISA titer against AMA1 domains1+2 >100,000. A negative correlation between log ELISA titer and day 11 cumulative parasitemia (Spearman rank r = -0.780, p value = 0.0001), further confirmed the relationship between antibody titer and protection. High titers of cross-strain inhibitory antibodies against AMA1 are therefore critical to confer solid protection, and the Aotus model can be used to down-select future AMA1 formulations, prior to advanced human trials.


Assuntos
Anticorpos Antiprotozoários/imunologia , Malária Falciparum/imunologia , Malária Falciparum/prevenção & controle , Proteínas de Protozoários/imunologia , Sequência de Aminoácidos , Animais , Formação de Anticorpos/imunologia , Especificidade de Anticorpos/imunologia , Aotus trivirgatus , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Imunoensaio , Vacinas Antimaláricas/química , Vacinas Antimaláricas/imunologia , Malária Falciparum/complicações , Dados de Sequência Molecular , Parasitemia/complicações , Parasitemia/imunologia , Plasmodium falciparum/crescimento & desenvolvimento , Plasmodium falciparum/imunologia , Dobramento de Proteína , Proteínas de Protozoários/química , Proteínas Recombinantes/química , Proteínas Recombinantes/imunologia , Análise de Sequência de Proteína , Titulometria , Vacinação
15.
Vector Borne Zoonotic Dis ; 9(5): 569-72, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19835479

RESUMO

William Collins, Ph.D., received his B.S. and M.Sc. degrees in entomology from Michigan State University. He completed his Ph.D. at Rutgers University in two years, just before being inducted into the Army to serve in the Korean War. He was assigned to Fort Detrick at the Biological Warfare Research Laboratories and after three years returned to Rutgers as an extension entomologist. He accepted a position in 1959 with the U.S. Public Health Service, with which he has worked for the last 50 years. In 1963, the Public Health Service laboratory moved to Atlanta and Dr. Collins' group began working with non-human primates following the discovery that monkey malarias were transmissible to humans. Parasites from monkeys or apes isolated in Asia, South America, and Africa were sent to the laboratory in Chamblee, Georgia, where they were adapted and transmitted to laboratory-maintained primates and their life cycles described and characterized. Transmissions to human volunteers were also attempted. In 1973, the laboratory operation was transferred to the CDC, and the emphasis changed from the study of monkey malaria in monkeys to that of human malaria in monkeys. During the last 25 to 30 years, different isolates of human malaria parasites have been adapted to New World monkeys to characterize the isolates for the development and testing of drugs and vaccines. Dr. Collins' task has been to identify and choose the best combination of vector-parasite-host combinations for testing each vaccine candidate. He has co-authored more than 450 manuscripts and has been awarded the U.S. Public Health Service Superior Service Award, The Joseph A. LaPrince Medal for Malariology from the American Society of Tropical Medicine and Hygiene, The Distinguished Service Award of the Department of Health and Human Services, the William Watson Medal of Excellence from the Centers for Disease Control and Prevention, and the Hoogstraal Medal from the American Committee of Medical Entomology.


Assuntos
Entomologia/história , Animais , Distinções e Prêmios , Ética em Pesquisa , História do Século XX , Humanos , Projetos de Pesquisa
16.
Am J Trop Med Hyg ; 81(3): 529-33, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19706927

RESUMO

Of 1,004 positive lots of mosquitoes fed on 229 humans infected with Plasmodium falciparum, 46.2% had 1-10 oocysts/(+)gut, 21.2% had 10-30 oocysts/(+)gut, 22.2% had 30-100 oocysts/(+)gut, and 10.4% had > 100 oocysts/(+) gut. The highest levels of infection occurred between 6 and 15 days after the peak in the asexual parasite count. Of 2,281 lots of Anopheles freeborni mosquitoes fed on splenectomized Aotus monkeys infected with the Santa Lucia strain of P. falciparum, 1,191 were infected (52.2%). The highest intensity infections ranged from 2.78 oocysts per positive gut in mosquitoes fed on Aotus vociferans to 6.08 oocysts per positive gut for those fed on A. lemurinus griseimembra to 10.4 oocysts per positive gut for those fed on A. nancymaae. The pattern of infection for mosquitoes fed on splenectomized Aotus monkeys was similar to that obtained by feeding on humans, but the intensity, based on oocyst/(+)gut, was much lower.


Assuntos
Anopheles/parasitologia , Aotidae/parasitologia , Insetos Vetores/parasitologia , Malária Falciparum/transmissão , Doenças dos Macacos/transmissão , Plasmodium falciparum/fisiologia , Animais , Interações Hospedeiro-Parasita , Humanos
17.
Am J Trop Med Hyg ; 80(5): 739-44, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19407117

RESUMO

Macaca mulatta monkeys infected with the Hackeri strain of Plasmodium fieldi had maximum parasite counts ranging from 1,300 to 301,320/microL. In 43 intact animals infected with the ABI strain, the maximum parasite counts ranged from 672 to 57,189/microL (median = 15,100/microL); in 46 splenectomized monkeys, the maximum parasite count ranged from 660 to 350,000/microL (median = 52,245/microL). Transmission through Anopheles dirus mosquitoes was obtained on 11 occasions with pre-patent periods of 9-14 days. Relapses occurred between two and eight times during a 1-year period. P. fieldi has potential for testing prophylactic and radical curative drugs.


Assuntos
Anopheles/parasitologia , Insetos Vetores/parasitologia , Macaca mulatta/parasitologia , Malária/parasitologia , Plasmodium/classificação , Animais , Antimaláricos/uso terapêutico , Modelos Animais de Doenças , Malária/tratamento farmacológico , Malária/transmissão , Oocistos/fisiologia , Fatores de Tempo
18.
Am J Trop Med Hyg ; 80(4): 536-40, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19346371

RESUMO

The Santa Lucia strain of Plasmodium falciparum was studied in 150 Aotus lemurinus griseimembra, 30 A. azarae boliviensis, 103 A. nancymaae, and 121 A. vociferans monkeys. All four of these splenectomized hosts supported the production of gametocytes infective to Anopheles freeborni mosquitoes. Transmission through sporozoites from An. freeborni, An. stephensi, An. maculatus, and An. albimanus mosquitoes was successful to all four species of Aotus on a total of 100 occasions with a median pre-patent period of 21 days. For the production of infective mosquitoes for vaccine challenge studies, A. l. griseimembra and A. vociferans were the most predictable hosts.


Assuntos
Aotidae/parasitologia , Malária Falciparum/parasitologia , Plasmodium falciparum/classificação , Animais , Anopheles/classificação , Anopheles/parasitologia , Aotidae/classificação , Pré-Escolar , Modelos Animais de Doenças , Feminino , Humanos , Insetos Vetores/classificação , Insetos Vetores/parasitologia , Malária Falciparum/transmissão , Plasmodium falciparum/fisiologia , Esplenectomia , Fatores de Tempo
20.
Am J Trop Med Hyg ; 80(2): 228-35, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19190218

RESUMO

A review is presented on studies conducted in New World monkeys and chimpanzees with the Salvador I strain of Plasmodium vivax. This isolate has been adapted to Aotus and Saimiri (squirrel) monkeys and developed as a model for the testing of antimalarial vaccines. After the injection of 10,000 sporozoites, the median prepatent period in S. boliviensis monkeys was 21.5 days. In 103 sporozoite-induced infections in splenectomized monkeys, the median maximum parasite count ranged from 2,139 to 202,368/microL, with a median maximum parasite count of 48,174/microL. Median maximum parasite counts in Aotus lemurinus griseimembra, A. nancymaae, A. azarae boliviensis, and A. vociferans monkeys were 19,902, 18,390, 21,420, and 18,210/microL, respectively and ranged from 124 to 156,000/microL. Mosquito infections were readily obtained in different species of Anopheles mosquitoes. The S. boliviensis monkey and Salvador I strain seems suitable for the testing of sporozoite and liver stage vaccines but not for blood-stage vaccines against P. vivax unless adapted further in spleen-intact Saimiri boliviensis monkeys.


Assuntos
Anopheles/parasitologia , Aotidae/parasitologia , Modelos Animais de Doenças , Malária Vivax/parasitologia , Plasmodium vivax/patogenicidade , Saimiri/parasitologia , Animais , Humanos , Fígado/parasitologia , Vacinas Antimaláricas/uso terapêutico , Malária Vivax/prevenção & controle , Plasmodium vivax/crescimento & desenvolvimento , Esporozoítos/crescimento & desenvolvimento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...